Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 9(4): 807-14, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20139729

ABSTRACT

Expression of Epstein-Barr Virus BZLF1, a key protein initiating the switch from latent to lytic infection, is known to cause cell growth arrest by accumulating p53 and p21(WAF1/CIP1) in epithelial cells, but its molecular mechanism remains elusive. We found here that the BZLF1 protein stimulates p53 binding to its recognition sequence. The BZLF1 accelerated the rate of p53-DNA complex formation through the interaction with p53 protein and also enhanced p53-specific transcription in vitro. Furthermore, p53 protein was found to bind to its target promoter regions specifically in the early stages of lytic replication. Overexpression of p53 at the early stages of lytic replication enhanced viral genome replication, supporting the idea that p53 plays an important role in the initiation steps of EBV replication. Taking the independent role of BZLF1 on p53 degradation into consideration, we propose that the BZLF1 protein regulates p53 and its target gene products in two distinctive manners; transient induction of p53 at the early stages for the initiation of viral productive replication and p53 degradation at the later stages for S-phase like environment preferable for viral replication.


Subject(s)
Herpesvirus 4, Human/genetics , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Virus Replication , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Viral , HeLa Cells , Humans , Transcriptional Activation
2.
Virology ; 398(2): 149-57, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20034650

ABSTRACT

We previously demonstrated that Bombyx mori nucleopolyhedrovirus (BmNPV) multiplication is restricted in permissive BmN-4 cells upon coinfection with Hyphantria cunea NPV (HycuNPV). Here, we show that HycuNPV-encoded hycu-ep32 gene is responsible for the restricted BmNPV multiplication in HycuNPV-coinfected BmN-4 cells. The only homologue for hycu-ep32 is in Orgyia pseudotsugata NPV. hycu-ep32 could encode a polypeptide of 312 amino acids, and it contains no characteristic domains or motifs to suggest its possible functions. hycu-ep32 is an early gene, and Hycu-EP32 expression reaches a maximum by 6 h postinfection. hycu-ep32-defective HycuNPV, vHycuDeltaep32, was generated, indicating that hycu-ep32 is nonessential in permissive SpIm cells. In BmN-4 cells, HycuNPV infection resulted in a severe global protein synthesis shutdown, while vHycuDeltaep32 did not cause any specific protein synthesis shutdown. These results indicate that the restriction of BmNPV multiplication by HycuNPV is caused by a global protein synthesis shutdown induced by hycu-ep32 upon coinfection with HycuNPV.


Subject(s)
Bombyx/virology , Genes, Viral/genetics , Moths/virology , Nucleopolyhedroviruses/genetics , Protein Biosynthesis/genetics , Virus Replication/genetics , Animals , Cell Line , Gene Expression Regulation, Viral/genetics , Genes, Viral/physiology , Immunoblotting , Nucleopolyhedroviruses/physiology , Protein Biosynthesis/physiology
3.
PLoS Pathog ; 5(7): e1000530, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19649319

ABSTRACT

p53-signaling is modulated by viruses to establish a host cellular environment advantageous for their propagation. The Epstein-Barr virus (EBV) lytic program induces phosphorylation of p53, which prevents interaction with MDM2. Here, we show that induction of EBV lytic program leads to degradation of p53 via an ubiquitin-proteasome pathway independent of MDM2. The BZLF1 protein directly functions as an adaptor component of the ECS (Elongin B/C-Cul2/5-SOCS-box protein) ubiquitin ligase complex targeting p53 for degradation. Intringuingly, C-terminal phosphorylation of p53 resulting from activated DNA damage response by viral lytic replication enhances its binding to BZLF1 protein. Purified BZLF1 protein-associated ECS could be shown to catalyze ubiquitination of phospho-mimetic p53 more efficiently than the wild-type in vitro. The compensation of p53 at middle and late stages of the lytic infection inhibits viral DNA replication and production during lytic infection, suggesting that the degradation of p53 is required for efficient viral propagation. Taken together, these findings demonstrate a role for the BZLF1 protein-associated ECS ligase complex in regulation of p53 phosphorylated by activated DNA damage signaling during viral lytic infection.


Subject(s)
Herpesvirus 4, Human/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cullin Proteins/metabolism , Herpesvirus 4, Human/growth & development , Humans , Molecular Sequence Data , Phosphorylation , Ubiquitination , Virus Replication
4.
Virology ; 388(1): 204-11, 2009 May 25.
Article in English | MEDLINE | ID: mdl-19375142

ABSTRACT

The Epstein-Barr virus (EBV) lytic program elicits ATM-dependent DNA damage response, resulting in phosphorylation of p53 at N-terminus, which prevents interaction with MDM2. Nevertheless, p53-downstream signaling is blocked. We found here that during the lytic infection p53 was actively degraded in a proteasome-dependent manner even with a reduced level of MDM2. BZLF1 protein enhanced the ubiquitination of p53 in SaOS-2 cells. The degradation of p53 was observed even in the presence of Nutlin-3, an inhibitor of p53-MDM2 interaction, and also in mouse embryo fibroblasts lacking mdm2 gene, indicating that the BZLF1 protein-induced degradation of p53 was independent of MDM2. Furthermore, Nutlin-3 increased the level of p53 in the latent phase of EBV infection but not in the lytic phase. Although p53 level is regulated by MDM2 in the latent phase, it might be mediated by the BZLF1 protein-associated E3 ubiquitin ligase in the lytic phase for efficient viral propagation.


Subject(s)
Gene Expression Regulation, Viral/physiology , Herpesvirus 4, Human/physiology , Proto-Oncogene Proteins c-mdm2/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Down-Regulation , Humans , Leupeptins , Mice , Trans-Activators/genetics , Transcription, Genetic
5.
J Virol ; 81(18): 9653-64, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17609267

ABSTRACT

The ataxia telangiectasia-mutated (ATM) protein, a member of the related phosphatidylinositol 3-like kinase family encoded by a gene responsible for the human genetic disorder ataxia telangiectasia, regulates cellular responses to DNA damage and viral infection. It has been previously reported that herpes simplex virus type 1 (HSV-1) infection induces activation of protein kinase activity of ATM and hyperphosphorylation of transcription factor, Sp1. We show that ATM is intimately involved in Sp1 hyperphosphorylation during HSV-1 infection rather than individual HSV-1-encoded protein kinases. In ATM-deficient cells or cells silenced for ATM expression by short hairpin RNA targeting, hyperphosphorylation of Sp1 was prevented even as HSV-1 infection progressed. Mutational analysis of putative ATM phosphorylation sites on Sp1 and immunoblot analysis with phosphopeptide-specific Sp1 antibodies clarified that at least Ser-56 and Ser-101 residues on Sp1 became phosphorylated upon HSV-1 infection. Serine-to-alanine mutations at both sites on Sp1 considerably abolished hyperphosphorylation of Sp1 upon infection. Although ATM phosphorylated Ser-101 but not Ser-56 on Sp1 in vitro, phosphorylation of Sp1 at both sites was not detected at all upon infection in ATM-deficient cells, suggesting that cellular kinase(s) activated by ATM could be involved in phosphorylation at Ser-56. Upon viral infection, Sp1-dependent transcription in ATM expression-silenced cells was almost the same as that in ATM-intact cells, suggesting that ATM-dependent phosphorylation of Sp1 might hardly affect its transcriptional activity during the HSV-1 infection. ATM-dependent Sp1 phosphorylation appears to be a global response to various DNA damage stress including viral DNA replication.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Protein Processing, Post-Translational/physiology , Protein Serine-Threonine Kinases/metabolism , Sp1 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Virus Replication/physiology , Amino Acid Substitution , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia/virology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , DNA Damage/genetics , DNA Replication/genetics , DNA, Viral/genetics , DNA, Viral/metabolism , DNA-Binding Proteins/genetics , Gene Silencing , HeLa Cells , Herpes Simplex/genetics , Herpesvirus 1, Human/genetics , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Sp1 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Viral Proteins/metabolism
6.
J Virol ; 80(20): 10064-72, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17005684

ABSTRACT

Induction of Epstein-Barr virus (EBV) lytic replication blocks chromosomal DNA replication notwithstanding an S-phase-like cellular environment with high cyclin-dependent kinase (CDK) activity. We report here that the phosphorylated form of MCM4, a subunit of the MCM complex essential for chromosomal DNA replication, increases with progression of lytic replication, Thr-19 and Thr-110 being CDK2/CDK1 targets whose phosphorylation inactivates MCM4-MCM6-MCM7 (MCM4-6-7) complex-associated DNA helicase. Expression of EBV-encoded protein kinase (EBV-PK) in HeLa cells caused phosphorylation of these sites on MCM4, leading to cell growth arrest. In vitro, the sites of MCM4 of the MCM4-6-7 hexamer were confirmed to be phosphorylated with EBV-PK, with the same loss of helicase activity as with CDK2/cyclin A. Introducing mutations in the N-terminal six Ser and Thr residues of MCM4 reduced the inhibition by CDK2/cyclin A, while EBV-PK inhibited the helicase activities of both wild-type and mutant MCM4-6-7 hexamers, probably since EBV-PK can phosphorylate MCM6 and another site(s) of MCM4 in addition to the N-terminal residues. Therefore, phosphorylation of the MCM complex by redundant actions of CDK and EBV-PK during lytic replication might provide one mechanism to block chromosomal DNA replication in the infected cells through inactivation of DNA unwinding by the MCM4-6-7 complex.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Helicases/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/physiology , Nuclear Proteins/metabolism , Virus Replication/physiology , Amino Acid Substitution , Cell Division , Cyclin A/metabolism , Cyclin-Dependent Kinase 2/metabolism , DNA/metabolism , HeLa Cells , Humans , Minichromosome Maintenance Complex Component 4 , Mutagenesis, Site-Directed , Mutation, Missense , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Processing, Post-Translational , Threonine/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
7.
J Gen Virol ; 87(Pt 9): 2549-2562, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16894193

ABSTRACT

The whole-genome sequence of the Hyphantria cunea nucleopolyhedrovirus (HycuNPV) was analysed. The entire nucleotide sequence of the HycuNPV genome was 132 959 bp long, with a G+C content of 45.1 mol%. A total of 148 open reading frames (ORFs) consisting of more than 50 aa were encoded by the genome. HycuNPV shares more than 122 ORFs with other lepidopteran group I NPVs, including Autographa californica MNPV, Bombyx mori NPV, Choristoneura fumiferana MNPV (CfMNPV), Choristoneura fumiferana defective NPV, Epiphyas postvittana MNPV and Orgyia pseudotsugata MNPV (OpMNPV). Six ORFs are identified as being unique to HycuNPV. Most of the HycuNPV ORFs showed higher similarity to CfMNPV and OpMNPV ORFs than to those of the other group I NPVs. HycuNPV encodes two conotoxin-like homologues (ctls), which are observed only in OpMNPV in group I NPVs. HycuNPV encodes three inhibitors of apoptosis (iaps), hycu-iap-1, hycu-iap-2 and hycu-iap-3, a feature that it shares only with CfMNPV. In addition, six homologous regions (hrs) are identified in the HycuNPV genome. These hrs are located in regions similar to those of the OpMNPV hrs, but different from most of the CfMNPV hrs. Based on the close phylogenetic relationship and conservation of group I NPV-specific genes, such as gp64, ie-2 and ptp-1, it is concluded that HycuNPV belongs to the group I NPVs and is most similar to CfMNPV or OpMNPV.


Subject(s)
Genome, Viral , Moths/virology , Nucleopolyhedroviruses/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Viral/genetics , Evolution, Molecular , Inhibitor of Apoptosis Proteins/genetics , Molecular Sequence Data , Multigene Family , Nucleopolyhedroviruses/classification , Nucleopolyhedroviruses/pathogenicity , Open Reading Frames , Phylogeny , Repetitive Sequences, Nucleic Acid , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Viral Proteins/genetics
8.
J Biol Chem ; 281(16): 11422-30, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16510450

ABSTRACT

The mismatch repair (MMR) system, highly conserved throughout evolution, corrects nucleotide mispairing that arise during cellular DNA replication. We report here that proliferating cell nuclear antigen (PCNA), the clamp loader complex (RF-C), and a series of MMR proteins like MSH-2, MSH-6, MLH1, and hPSM2 can be assembled to Epstein-Barr virus replication compartments, the sites of viral DNA synthesis. Levels of the DNA-bound form of PCNA increased with progression of viral productive replication. Bromodeoxyuridine-labeled chromatin immunodepletion analyses confirmed that PCNA is loaded onto newly synthesized viral DNA as well as BALF2 and BMRF1 viral proteins during lytic replication. Furthermore, the anti-PCNA, -MSH2, -MSH3, or -MSH6 antibodies could immunoprecipitate BMRF1 replication protein probably via the viral DNA genome. PCNA loading might trigger transfer of a series of host MMR proteins to the sites of viral DNA synthesis. The MMR factors might function for the repair of mismatches that arise during viral replication or act to inhibit recombination between moderately divergent (homologous) sequences.


Subject(s)
Base Pair Mismatch , DNA Repair , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/physiology , Virus Replication , Adaptor Proteins, Signal Transducing , Adenosine Triphosphatases/metabolism , Animals , Antigens, Viral/metabolism , Apoptosis , Bromodeoxyuridine/pharmacology , Carrier Proteins/metabolism , Cell Cycle , Cell Line , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA/metabolism , DNA Damage , DNA Repair Enzymes/metabolism , DNA Replication , DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Epstein-Barr Virus Nuclear Antigens/chemistry , Genome, Viral , Humans , Immunoblotting , Immunoprecipitation , Mice , Microscopy, Fluorescence , Mismatch Repair Endonuclease PMS2 , MutL Protein Homolog 1 , MutS Homolog 2 Protein/metabolism , Nuclear Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Recombination, Genetic , Subcellular Fractions/metabolism , Time Factors , Viral Proteins/metabolism
9.
J Virol ; 79(15): 9597-607, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014922

ABSTRACT

We previously demonstrated that the major immediate early (MIE) proximal enhancer containing one GC box and the TATA box containing promoter are minimal elements required for transcription and viral replication in human fibroblast cells (H. Isomura, T. Tsurumi, M. F. Stinski, J. Virol. 78:12788-12799, 2004). After infection, the level of Sp1 increased while Sp3 remained constant. Here we report that either Sp1 or Sp3 transcription factors bind to the GC boxes located at approximately positions -55 and -75 relative to the transcription start site (+1). Both the Sp1 and Sp3 binding sites have a positive and synergistic effect on the human cytomegalovirus (HCMV) major immediate-early (MIE) promoter. There was little to no change in MIE transcription or viral replication for recombinant viruses with one or the other Sp1 or Sp3 binding site mutated. In contrast, mutation of both the Sp1 and Sp3 binding sites caused inefficient MIE transcription and viral replication. These data indicate that the Sp1 and Sp3 binding sites have a significant role in HCMV replication in human fibroblast cells.


Subject(s)
Binding Sites/physiology , Cytomegalovirus/genetics , Genes, Immediate-Early/physiology , Promoter Regions, Genetic/physiology , Transcription Factors/physiology , Fibroblasts , Gene Expression Regulation, Viral , Humans , Protein Binding , Transcription Factors/metabolism , Virus Replication
10.
J Biol Chem ; 280(34): 30336-41, 2005 Aug 26.
Article in English | MEDLINE | ID: mdl-15964848

ABSTRACT

Eukaryotic cells are equipped with machinery to monitor and repair damaged DNA. Herpes simplex virus (HSV) DNA replication occurs at discrete sites in nuclei, the replication compartment, where viral replication proteins cluster and synthesize a large amount of viral DNA. In the present study, HSV infection was found to elicit a cellular DNA damage response, with activation of the ataxia-telangiectasia-mutated (ATM) signal transduction pathway, as observed by autophosphorylation of ATM and phosphorylation of multiple downstream targets including Nbs1, Chk2, and p53, while infection with a UV-inactivated virus or with a replication-defective virus did not. Activated ATM and the DNA damage sensor MRN complex composed of Mre11, Rad50, and Nbs1 were recruited and retained at sites of viral DNA replication, probably recognizing newly synthesized viral DNAs as abnormal DNA structures. These events were not observed in ATM-deficient cells, indicating ATM dependence. In Nbs1-deficient cells, HSV infection induced an ATM DNA damage response that was delayed, suggesting a functional MRN complex requirement for efficient ATM activation. However, ATM silencing had no effect on viral replication in 293T cells. Our data open up an interesting question of how the virus is able to complete its replication, although host cells activate ATM checkpoint signaling in response to the HSV infection.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Herpesviridae Infections/metabolism , Protein Serine-Threonine Kinases/metabolism , Simplexvirus/metabolism , Tumor Suppressor Proteins/metabolism , Acid Anhydride Hydrolases , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line , Checkpoint Kinase 2 , Chlorocebus aethiops , DNA Damage , DNA Repair Enzymes/metabolism , DNA, Viral , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Gene Silencing , Humans , Immunoblotting , In Situ Hybridization, Fluorescence , Kinetics , MRE11 Homologue Protein , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Signal Transduction , Time Factors , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism , Ultraviolet Rays , Up-Regulation , Vero Cells
11.
J Virol ; 79(6): 3409-18, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731235

ABSTRACT

Epstein-Barr virus (EBV) productive DNA replication occurs at discrete sites, called replication compartments, in nuclei. In this study we performed comprehensive analyses of the architecture of the replication compartments. The BZLF1 oriLyt binding proteins showed a fine, diffuse pattern of distribution throughout the nuclei at immediate-early stages of induction and then became associated with the replicating EBV genome in the replication compartments during lytic infection. The BMRF1 polymerase (Pol) processivity factor showed a homogenous, not dot-like, distribution in the replication compartments, which completely coincided with the newly synthesized viral DNA. Inhibition of viral DNA replication with phosphonoacetic acid, a viral DNA Pol inhibitor, eliminated the DNA-bound form of the BMRF1 protein, although the protein was sufficiently expressed in the cells. These observations together with the findings that almost all abundantly expressed BMRF1 proteins existed in the DNA-bound form suggest that the BMRF1 proteins not only act at viral replication forks as Pol processive factors but also widely distribute on newly replicated EBV genomic DNA. In contrast, the BALF5 Pol catalytic protein, the BALF2 single-stranded-DNA binding protein, and the BBLF2/3 protein, a component of the helicase-primase complex, were colocalized as distinct dots distributed within replication compartments, representing viral replication factories. Whereas cellular replication factories are constructed based on nonchromatin nuclear structures and nuclear matrix, viral replication factories were easily solubilized by DNase I treatment. Thus, compared with cellular DNA replication, EBV lytic DNA replication factories would be simpler so that construction of the replication domain would be more relaxed.


Subject(s)
Cell Nucleus/chemistry , Herpesvirus 4, Human/physiology , Virus Replication , Antigens, Viral/metabolism , DNA Replication , DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/metabolism , Deoxyribonuclease I/metabolism , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Phosphonoacetic Acid/pharmacology , Trans-Activators/metabolism , Viral Proteins/metabolism
12.
J Biol Chem ; 280(9): 8156-63, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15611093

ABSTRACT

When exposed to genotoxic stress, eukaryotic cells demonstrate a DNA damage response with delay or arrest of cell-cycle progression, providing time for DNA repair. Induction of the Epstein-Barr virus (EBV) lytic program elicited a cellular DNA damage response, with activation of the ataxia telangiectasia-mutated (ATM) signal transduction pathway. Activation of the ATM-Rad3-related (ATR) replication checkpoint pathway, in contrast, was minimal. The DNA damage sensor Mre11-Rad50-Nbs1 (MRN) complex and phosphorylated ATM were recruited and retained in viral replication compartments, recognizing newly synthesized viral DNAs as abnormal DNA structures. Phosphorylated p53 also became concentrated in replication compartments and physically interacted with viral BZLF1 protein. Despite the activation of ATM checkpoint signaling, p53-downstream signaling was blocked, with rather high S-phase CDK activity associated with progression of lytic infection. Therefore, although host cells activate ATM checkpoint signaling with response to the lytic viral DNA synthesis, the virus can skillfully evade this host checkpoint security system and actively promote an S-phase-like environment advantageous for viral lytic replication.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Bromodeoxyuridine/pharmacology , Cell Line , Cell Line, Tumor , DNA Damage , Humans , Immunoblotting , Immunoprecipitation , In Situ Hybridization, Fluorescence , MRE11 Homologue Protein , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Protein Kinases/metabolism , S Phase , Serine/chemistry , Signal Transduction , Time Factors , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...